Tadpole tail regeneration in Xenopus

Biochemical Society Transactions

www.biochemsoctrans.org

Yaoyao Chen*†, Nick R. Love*‡ and Enrique Amaya*1 *The Healing Foundation Centre, Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester M13 9PT, U.K. †Wellcome Trust–Medical Research Council Cambridge Stem Cell Institute, Tennis Court Road, Cambridge CB2 1QR, U.K. ‡RIKEN Center for Developmental Biology, 650-0047 Kobe, Japan

Abstract Some organisms have a remarkable ability to heal wounds without scars and to regenerate complex tissues following injury. By gaining a more complete understanding of the biological mechanisms that promote scar-free healing and tissue regeneration, it is hoped that novel treatments that can enhance the healing and regenerative capacity of human patients can be found. In the present article, we briefly examine the genetic, molecular and cellular mechanisms underlying the regeneration of the Xenopus tadpole tail.

Introduction Although mammals have relatively poor capacities to heal wounds and regenerate complex tissues, many organisms across the animal kingdom have remarkably good abilities to regenerate body parts following traumatic injuries. In these cases, the regeneration programme must be very robust, as no two injuries are ever the same. Successful regeneration relies on a co-ordinated orchestration of numerous processes, such as gene activation, signalling, cell proliferation, morphogenesis, tissue patterning and differentiation. How these proceed in a synchronized way during regeneration (and cease when regeneration is finished) is not very well understood. Ultimately, it is hoped that a better understanding of these aspects of regeneration will facilitate the development of improved and more efficient healing and regenerative therapies following traumatic injuries [1]. Investigations using invertebrate and vertebrate model organisms have greatly contributed to our understanding of the biology of regeneration [2,3]. Among vertebrate organisms, urodele amphibians are of particular note, as they have been reported to completely regenerate their arms, legs, tails, eye lens and jaw portions following removal [4,5]. In the present review, we examine another well-studied model of vertebrate regeneration: the anuran amphibian Xenopus, which is capable of fully regenerating its tail Key words: metabolism, reactive oxygen species, tissue regeneration, wound healing, Xenopus. Abbreviations: FGF, fibroblast growth factor; HIF, hypoxia-inducible factor; Ldha, lactate dehydrogenase A; Nrx, nucleoredoxin; Pdk1, pyruvate dehydrogenase kinase 1; PEP, phosphoenolpyruvate; PK, pyruvate kinase; ROS, reactive oxygen species; Slc, solute carrier. 1

To whom correspondence manchester.ac.uk).

should

be

addressed

(email

Biochem. Soc. Trans. (2014) 42, 617–623; doi:10.1042/BST20140061

enrique.amaya@

following amputation throughout its larval stage. For at least 60 years, scientists have examined the regeneration of the Xenopus tadpole tail [6–9], and thus the literature pertaining to this model has been reviewed previously [10–12]. In the present review, we briefly examine the genetic, molecular and cellular mechanisms underlying the regeneration of the Xenopus tadpole tail before focusing on recent evidence concerning the metabolic regulation of the regenerative process.

The Xenopus tadpole tail regeneration model

Biochemical Society Annual Symposium No. 81

Biochemical Determinants of Tissue Regeneration

As an experimental model, aquatic Xenopus frogs have numerous advantages [13]: they are relatively inexpensive to house; tadpoles can be generated in very large numbers, by either induction of natural matings, following the administration of hormones, or by in vitro fertilizations at any time of the year; the Xenopus model has extensive genomic resources, including the published genome of Xenopus tropicalis [14] and there exist over a million ESTs and thousands of gene clones, which expedite gainof-function and in situ hybridization studies [15–17]. Moreover, Xenopus transgenic technology permits tissuespecific, inducible and binary transgene expression systems that facilitate tail-regeneration experiments [18–21]. Finally, the recent development of genome-editing techniques will greatly enhance our ability to study the genetic basis of regeneration in this system [22]. The tadpole tail is a complex appendage, containing many axial tissues that are also found in humans (e.g. the spinal cord  C The

Authors Journal compilation

 C 2014

Biochemical Society

617

618

Biochemical Society Transactions (2014) Volume 42, part 3

and dorsal aorta). Moreover, the Xenopus tadpole tail contains a notochord, a dense network of neurons, skeletal and smooth muscle, lymphatic vasculature, and an epidermis dotted with black pigmented melanophores. All of these tissues regenerate and return to a fully functional tail appendage within 1 week of amputation.

The early, intermediate and late phases of Xenopus tadpole tail regeneration Tadpole tail regeneration proceeds through three phases: early, intermediate and late [23,24] (Figure 1). In the early phase (within the first 6 h of amputation), the site of injury undergoes scarless wound healing. This stage is also associated with an acute inflammatory response [24,25], which can be assayed using the histological stain Sudan Black B, which strongly reacts with neutrophil granules in inflammatory cells, [26] or by in situ hybridization using marker genes, such as Mmp7 (matrix metalloproteinase 7) or Mpo (myeloperoxidase) [27,28]. The inflammatory response can also be imaged using time-lapse microscopy on transgenic lines, which label myeloid cells with GFP [29,30]. At the injury site, inflammatory cells help to prevent microbial infection and participate in other phagocytic and nonphagocytic duties. Several other activities have been shown to be critical during the early phase of tail regeneration. For example, during the first 6 h of tail amputation, the V-ATPase (vacuolar ATPase) H + pump is activated and results in an electrically polarized injured tail [31], which is required for proper regeneration. In addition, during this phase of regeneration, a moderate number of cells at the injured site undergo apoptosis, and this is required for tail regeneration to proceed normally [32]. Given the evidence that amputation-induced apoptotic cells can release growth factors during Hydra regeneration [33], it is possible that these apoptotic cells at the distal part of the injured tail are also a source of proregenerative signals during Xenopus tail regeneration. The next phase of regeneration, the intermediate phase (∼24–36 h after amputation), is classified by the formation of regenerative tissue distal to the wound site. Although this nascent regenerative tissue is sometimes referred to as the ‘blastema’, we (and others) prefer to refer to this tissue as the ‘regenerative bud’ [34]. The regenerative bud is highly dynamic and can take various shapes during regeneration. Although the exact role of the regenerative bud is not entirely understood, its formation is generally thought to be required for tail regeneration. In addition, grafting experiments have shown that the cells of the regenerative bud do not appear to contribute to or ‘seed’ the regenerating muscle, spinal cord or notochord tissues [35]. However, the regenerative bud clearly possesses an intrinsic patterning ability and can promote tissue outgrowth: transplanting a genetically labelled regenerative bud into the head region of a tadpole will cause an ectopic tail to grow, and the ectopic tail’s epidermis retains the genetic label [36].  C The

C 2014 Biochemical Society Authors Journal compilation 

Following the formation of the regenerative bud, the overt regrowth of the tail appendage begins during the late phase of tadpole tail regeneration. During this time, the vascular, neuronal, notochord and muscle tissues all repopulate the new tail from ∼24–36 h after amputation until ∼5–9 days after amputation [30]. As one might expect, successful tail regeneration requires the activity of many growth factors, including the BMP (bone morphogenetic protein), Notch, Wnt, FGF (fibroblast growth factor) and TGFβ (transforming growth factor β) [20,37,38]. Given the interest in spinal cord regeneration following human injury, the mechanisms of tadpole spinal cord regeneration are particularly intriguing. Grafting experiments using GFP-expressing transgenic tadpoles have elegantly shown that the injured spinal cord (and notochord) provides the cellular source for their regenerative portions [35]. The presence of the spinal cord is essential for regeneration, as its removal before amputation will severely compromise the regenerative response [39]. Tail muscle regenerates from a reservoir of resident muscle stem cells, called satellite cells [35]. It is now appreciated that the transcription factor Pax7 is essential for satellite cells to be able to mount a regenerative response [40]. More recent studies have used fluorophores to track the dynamic behaviour of myofibrils during tail muscle regeneration, suggesting that injured tail muscle fibres show some signs of dedifferentiation following amputation [41]. The regeneration of the tadpole tail vasculature has only been minimally studied [24]. However, there currently exist several transgenic lines in both Xenopus laevis [42] and X. tropicalis [24] that can be used to image the regeneration of the vasculature in vivo [30]. The availability of these lines should facilitate the study of tadpole vascular regeneration. It is expected that the vasculature is essential for providing oxygen and nourishment to the regenerating tail.

Gene expression during tail regeneration Tail amputation and the subsequent regeneration programme result in the alteration of gene expression level in thousands of genes during regeneration [23,24]. The change in gene expression following amputation is likely to depend on remodelling of the epigenetic landscape of the genome, and, consistent with this prediction, HDAC (histone deacetylase) activity has recently been shown to be essential for amputation-induced change in mRNA transcription [34,43]. One fundamental question regarding tail regeneration is whether the regeneration gene expression programme is similar to that seen during the initial tail developmental programme. This question of ‘recapitulation’ was addressed by Mochii and colleagues, who found that several developmental genes (i.e. chordin, noggin, Xshh and delta1) were expressed during development but not during regeneration [44]. Moreover, in a separate study, it was also found that the expression of abdominal B-type Hox genes hoxc10, hoxa13 and hoxd13 were not the same during tail embryonic development and regeneration [45].

Biochemical Society Annual Symposium No. 81: Biochemical Determinants of Tissue Regeneration

Figure 1 Overview of events that occur during tail regeneration Tail regeneration proceeds through three phases. Soon after injury, the amputation site begins to produce ROS, and this production is sustained through the regeneration process. During regeneration, a large cohort of genes change in expression and many of these are associated with signalling and metabolism (depicted in the close-up inset from the 24 h after amputation time point). Many of these changes would be predicted to lead to increased anabolic pathways and attenuation of catabolic pathways, thereby facilitating tissue growth and regeneration. G6P, glucose 6-phosphate; g6pd, glucose-6-phosphate dehydrogenase; PPP, pentose phosphate pathway.

 C The

C 2014 Biochemical Society Authors Journal compilation 

619

620

Biochemical Society Transactions (2014) Volume 42, part 3

However, other genes, such as Fgf20, clearly reinitiate their developmental expression patterns during tail regeneration [17,38]. Thus regeneration recapitulates some gene expression patterns exhibited during tail development, but not all genes do so.

The role of reactive oxygen species during tail regeneration We performed a microarray screen aimed at examining the changes in gene expression during Xenopus tail regeneration, uncovering a number of co-ordinately up-regulated genes involved in the production of ROS (reactive oxygen species) such as H2 O2 [24]. Indeed, by using an oxidativesensitive ratiometric reporter fluorophore (HyPerYFP), we were able to confirm that tail amputation and the subsequent regenerative response was associated with a significant increase in the production of intracellular ROS levels [46] (Figure 1). By monitoring the entire process of tail regeneration, we found that this oxidative change, which is equivalent to 50–200 μM H2 O2 , lasts for at least several days before it eventually wanes. Interestingly, this amputation-induced ROS not only precedes the recruitment of inflammatory cells, but also functions independently of inflammatory cells. H2 O2 and other ROS, originally thought to cause stress and apoptosis, now have been increasingly appreciated as important regulators of various cellular processes, including cell metabolism, motility, proliferation and signalling [47,48]. Indeed, we found that decreasing ROS levels during tail regeneration via chemical inhibitors or gene-knockdown strategies, especially during the first 72 h after amputation, resulted in sluggish cell proliferation and impaired tail regeneration [46]. To better understand the potential mechanisms that might be affected by ROS during the process of tail regeneration, we asked whether Wnt/β-catenin and/or FGF signalling acted downstream of ROS production, as both of these growth factors had been shown previously to be critical for tail regeneration [38]. By using a X. tropicalis Wnt/β-catenin reporter line [49], we observed a sustained activation of Wnt/β-catenin signalling from 24 h after amputation and we found that inhibiting ROS using chemical compounds resulted in a significant decrease in the activation of Wnt/βcatenin signalling. It has been reported that Wnt/β-catenin signalling can be modulated by H2 O2 through a small redox-sensitive protein, Nrx (nucleoredoxin) [50]. Further investigation is needed to assess whether Nrx also mediates the changes in Wnt/β-catenin signalling activity caused by increased ROS levels during tail regeneration. FGF signalling has also been shown to act downstream of Wnt/β-catenin signalling during vertebrate appendage regeneration, as its inhibition blocks proliferation of blastemal cells during zebrafish caudal fin regeneration [51]. Among all of the genes that encode FGF ligands, we and others found that Fgf20 was among the most highly upregulated Fgf genes found during both X. laevis and X. tropicalis tail regeneration [24,38]. We and others also found  C The

C 2014 Biochemical Society Authors Journal compilation 

that inhibiting the expression of Fgf20 or its activity by either chemical or genetic means results in impaired tail regeneration [38,46]. Notably, the activation of Fgf20 gene expression during tail regeneration was significantly impaired when we treated tadpoles with ROS inhibitors, suggesting that the production of ROS following tail amputation is important for proper FGF signalling during regeneration [46]. Taken together, we speculate that the amputationinduced ROS facilitates the activation of Wnt/β-catenin and FGF signalling, both of which are crucial for tadpole tail regeneration. However, apart from modulating these two signalling pathways, ROS also affect other aspects of cellular physiology, such as cellular metabolism, thereby potentially creating a permissive environment for tissues to repair and regenerate.

Towards a greater understanding of the regulation and role of cellular metabolism during regeneration The metabolism of a living homoeostatic organism is well balanced between catabolic and anabolic reactions. However, under circumstances that require tissue regrowth, a higher rate of anabolism is intuitively preferred to meet the demands of new cellular components. In fact, scientists noticed decades ago that the citric acid cycle was not favoured during preblastemic and blastemic phases of tissue regeneration, and that glucose metabolism was tuned towards glycolysis and the PPP (pentose phosphate pathway) to provide intermediate metabolites to fuel anabolic processes, as well as to create an acidic local environment which was thought to be essential for many processes of regeneration [52]. However, it has remained unclear exactly how glucose metabolism is modulated during regeneration, since the earlier studies were restrained by the lack of decoded genome information and large-scale ‘-omic’ approaches. However, now with the published genome resources of X. tropicalis [14] and X. laevis, our genome-wide microarray analysis of X. tropicalis tail regeneration, as well as other large-scale genetic analysis of Xenopus tissue repair [23,53–55], we are better able to incorporate these important genetic tools to gain more insight into our understanding of the regulation and role of metabolism during tissue regeneration. We found that the expression of a large number of metabolic genes was greatly altered during the early, intermediate and late phases of tail regeneration [24] (Figure 1). When we specifically looked at genes related to glucose metabolism, we found that the expression of genes that stimulate glucose intake, such as those encoding leptin and proinsulin, and genes encoding subunits of glucose transporters, such as Slc2a3 (solute carrier 2 A3), were significantly induced after tail amputation. These data strongly suggest that regenerating tadpole tails actively increase glucose consumption. Meanwhile, our gene analysis also revealed that mRNA levels of Hif1a (hypoxia-inducible factor 1α), a gene known to reduce mitochondrial activity [56], was increased during

Biochemical Society Annual Symposium No. 81: Biochemical Determinants of Tissue Regeneration

tail regeneration [24]. Moreover, it is likely that the HIF1α protein is also stabilized at the post-translational level as amputation cuts down the blood supply to the injured area and creates a hypoxic environment {a condition which abrogates the prolyl hydroxylation of HIF1α and allows it to escape recognition by the pVHL (von Hippel–Lindau protein) ubiquitin ligase complex for degradation [57]}. Notably, intracellular ROS have also been reported to inhibit the catalytic activity of the prolyl hydroxylase domain and therefore negatively regulate HIF1α degradation [58– 60]. As a result, HIF1α accumulates in the nucleus and transactivates HIF-responsive genes, including many key metabolic genes, such as Pdk1, Ldha and Slc2a1 [56,61– 64]. These metabolic genes may have important roles in shaping the metabolic environment during tissue repair: Pdk1 encodes pyruvate dehydrogenase kinase 1 which inhibits the conversion of pyruvate into acetyl-CoA by inactivating pyruvate dehydrogenase; Ldha encodes lactate dehydrogenase A which catalyses the conversion of pyruvate into lactate; and Slc2a1 encodes a major glucose transporter, GLUT1, which can further enhance the glucose intake. Thus these genes may act synergistically to promote glycolysis and restrain carbohydrates from entering the tricarboxylic acid cycle [65–67]. Another important checkpoint that controls the balance between glycolysis and oxidative phosphorylation is the conversion of PEP (phosphoenolpyruvate) into pyruvate. The enzyme which regulates this rate-limiting step has two splice isoforms, PK (pyruvate kinase) M1 and M2 [68]. Although it is yet to be tested which isoform is expressed during tadpole tail regeneration, it was suggested that the expression of PKM2 can be induced by HIF1α through a positive-feedback loop [69], therefore PKM2 may indeed be the major form of PKMs expressed in the hypoxic tail. Unlike PKM1, which is constitutively active, PKM2 can switch between an active tetrameric and an inactive dimeric form [70], and is susceptible to inhibition by growth factor signalling and ROS oxidation [71,72]. Thus we speculate that the PKM2-mediated inefficient conversion of PEP also facilitates the metabolic shift towards glycolysis during tail regrowth. Altogether, changes in gene expression, growth factor signalling and ROS co-operate to reprogramme metabolism during tail regeneration to meet the synthetic demands of building new biomass during appendage regeneration. A more complete understanding of tissue regeneration will include insights into whether modulating metabolic programming may improve tissue regeneration outcomes in other animal models that are less competent to regenerate, such as mammals.

Funding This work was supported by the Wellcome Trust [grant number WT082450MA], the Healing Foundation and the National Science Foundation.

References 1 Gurtner, G.C., Werner, S., Barrandon, Y. and Longaker, M.T. (2008) Wound repair and regeneration. Nature 453, 314–321 CrossRef PubMed 2 Brockes, J.P. and Kumar, A. (2008) Comparative aspects of animal regeneration. Annu. Rev. Cell Dev. Biol. 24, 525–549 CrossRef PubMed 3 Sanchez Alvarado, A. and Tsonis, P.A. (2006) Bridging the regeneration gap: genetic insights from diverse animal models. Nat. Rev. Genet. 7, 873–884 CrossRef PubMed 4 Brockes, J.P. and Kumar, A. (2005) Appendage regeneration in adult vertebrates and implications for regenerative medicine. Science 310, 1919–1923 CrossRef PubMed 5 Kragl, M. and Tanaka, E.M. (2009) Axolotl (Ambystoma mexicanum) limb and tail amputation. Cold Spring Harb. Protoc. 2009, pdb.prot5267 PubMed 6 Pestalozzi, D. (1953) The effect of roentgen irradiation of varying dosage on the regenerative power of the tail of Xenopus larvae. Oncologia 6, 161–190 CrossRef PubMed 7 Roguski, H. (1953) Regeneration of the tail of tadpole Xenopus laevis. Folia Biol. 1, 7–22 8 Roguski, H. (1953) Further studies on regeneration of the tail of tadpole Xenopus laevis. Folia Biol. 1, 277–285 9 von Hahn, H. (1959) Catalase activity in the regenerating tail tip of Xenopus larvae and the effect of 3-amino-1,2,4-triazole. Experientia 15, 379–380 CrossRef PubMed 10 Beck, C.W., Izpisua Belmonte, J.C. and Christen, B. (2009) Beyond early development: Xenopus as an emerging model for the study of regenerative mechanisms. Dev. Dyn. 238, 1226–1248 CrossRef PubMed 11 Slack, J.M., Lin, G. and Chen, Y. (2008) The Xenopus tadpole: a new model for regeneration research. Cell. Mol. Life Sci. 65, 54–63 CrossRef PubMed 12 Tseng, A.S. and Levin, M. (2008) Tail regeneration in Xenopus laevis as a model for understanding tissue repair. J. Dent. Res. 87, 806–816 CrossRef PubMed 13 Amaya, E. (2005) Xenomics. Genome Res. 15, 1683–1691 CrossRef PubMed 14 Hellsten, U., Harland, R.M., Gilchrist, M.J., Hendrix, D., Jurka, J., Kapitonov, V., Ovcharenko, I., Putnam, N.H., Shu, S., Taher, L. et al. (2010) The genome of the Western clawed frog Xenopus tropicalis. Science 328, 633–636 CrossRef PubMed 15 Gilchrist, M.J., Zorn, A.M., Voigt, J., Smith, J.C., Papalopulu, N. and Amaya, E. (2004) Defining a large set of full-length clones from a Xenopus tropicalis EST project. Dev. Biol. 271, 498–516 CrossRef PubMed 16 Chen, J.A., Voigt, J., Gilchrist, M., Papalopulu, N. and Amaya, E. (2005) Identification of novel genes affecting mesoderm formation and morphogenesis through an enhanced large scale functional screen in Xenopus. Mech. Dev. 122, 307–331 CrossRef PubMed 17 Lea, R., Papalopulu, N., Amaya, E. and Dorey, K. (2009) Temporal and spatial expression of FGF ligands and receptors during Xenopus development. Dev. Dyn. 238, 1467–1479 CrossRef PubMed 18 Kroll, K.L. and Amaya, E. (1996) Transgenic Xenopus embryos from sperm nuclear transplantations reveal FGF signaling requirements during gastrulation. Development 122, 3173–3183 PubMed 19 Love, N.R., Thuret, R., Chen, Y., Ishibashi, S., Sabherwal, N., Paredes, R., Alves-Silva, J., Dorey, K., Noble, A.M., Guille, M.J. et al. (2011) pTransgenesis: a cross-species, modular transgenesis resource. Development 138, 5451–5458 CrossRef PubMed 20 Beck, C.W., Christen, B. and Slack, J.M. (2003) Molecular pathways needed for regeneration of spinal cord and muscle in a vertebrate. Dev. Cell 5, 429–439 CrossRef PubMed 21 Beck, C.W., Christen, B., Barker, D. and Slack, J.M. (2006) Temporal requirement for bone morphogenetic proteins in regeneration of the tail and limb of Xenopus tadpoles. Mech. Dev. 123, 674–688 CrossRef PubMed 22 Ishibashi, S., Cliffe, R. and Amaya, E. (2012) Highly efficient bi-allelic mutation rates using TALENs in Xenopus tropicalis. Biol. Open 1, 1273–1276 CrossRef PubMed 23 Tazaki, A., Kitayama, A., Terasaka, C., Watanabe, K., Ueno, N. and Mochii, M. (2005) Macroarray-based analysis of tail regeneration in Xenopus laevis larvae. Dev. Dyn. 233, 1394–1404 CrossRef PubMed  C The

C 2014 Biochemical Society Authors Journal compilation 

621

622

Biochemical Society Transactions (2014) Volume 42, part 3

24 Love, N.R., Chen, Y., Bonev, B., Gilchrist, M.J., Fairclough, L., Lea, R., Mohun, T.J., Paredes, R., Zeef, L.A. and Amaya, E. (2011) Genome-wide analysis of gene expression during Xenopus tropicalis tadpole tail regeneration. BMC Dev. Biol. 11, 70 CrossRef PubMed 25 Fukazawa, T., Naora, Y., Kunieda, T. and Kubo, T. (2009) Suppression of the immune response potentiates tadpole tail regeneration during the refractory period. Development 136, 2323–2327 CrossRef PubMed 26 Sheehan, H.L. and Storey, G.W. (1947) An improved method of staining leucocyte granules with Sudan Black B. J. Pathol. Bacteriol. 59, 336 CrossRef PubMed 27 Harrison, M., Abu-Elmagd, M., Grocott, T., Yates, C., Gavrilovic, J. and Wheeler, G.N. (2004) Matrix metalloproteinase genes in Xenopus development. Dev. Dyn. 231, 214–220 CrossRef PubMed 28 Chen, Y., Costa, R.M., Love, N.R., Soto, X., Roth, M., Paredes, R. and Amaya, E. (2009) C/EBPα initiates primitive myelopoiesis in pluripotent embryonic cells. Blood 114, 40–48 CrossRef PubMed 29 Smith, S.J., Kotecha, S., Towers, N., Latinkic, B.V. and Mohun, T.J. (2002) XPOX2-peroxidase expression and the XLURP-1 promoter reveal the site of embryonic myeloid cell development in Xenopus. Mech. Dev. 117, 173–186 CrossRef PubMed 30 Love, N.R., Ziegler, M., Chen, Y. and Amaya, E. (2014) Carbohydrate metabolism during vertebrate appendage regeneration: what is its role? How is it regulated? A postulation that regenerating vertebrate appendages facilitate glycolytic and pentose phosphate pathways to fuel macromolecule biosynthesis. BioEssays 36, 27–33 CrossRef PubMed 31 Adams, D.S., Masi, A. and Levin, M. (2007) H + pump-dependent changes in membrane voltage are an early mechanism necessary and sufficient to induce Xenopus tail regeneration. Development 134, 1323–1335 CrossRef PubMed 32 Tseng, A.S., Adams, D.S., Qiu, D., Koustubhan, P. and Levin, M. (2007) Apoptosis is required during early stages of tail regeneration in Xenopus laevis. Dev. Biol. 301, 62–69 CrossRef PubMed 33 Galliot, B. and Chera, S. (2010) The Hydra model: disclosing an apoptosis-driven generator of Wnt-based regeneration. Trends Cell Biol. 20, 514–523 CrossRef PubMed 34 Tseng, A.S., Carneiro, K., Lemire, J.M. and Levin, M. (2011) HDAC activity is required during Xenopus tail regeneration. PLoS ONE 6, e26382 CrossRef PubMed 35 Gargioli, C. and Slack, J.M. (2004) Cell lineage tracing during Xenopus tail regeneration. Development 131, 2669–2679 CrossRef PubMed 36 Waldner, C., Roose, M. and Ryffel, G.U. (2009) Red fluorescent Xenopus laevis: a new tool for grafting analysis. BMC Dev. Biol. 9, 37 CrossRef PubMed 37 Ho, D.M. and Whitman, M. (2008) TGF-β signaling is required for multiple processes during Xenopus tail regeneration. Dev. Biol. 315, 203–216 CrossRef PubMed 38 Lin, G. and Slack, J.M. (2008) Requirement for Wnt and FGF signaling in Xenopus tadpole tail regeneration. Dev. Biol. 316, 323–335 CrossRef PubMed 39 Taniguchi, Y., Sugiura, T., Tazaki, A., Watanabe, K. and Mochii, M. (2008) Spinal cord is required for proper regeneration of the tail in Xenopus tadpoles. Dev. Growth Differ. 50, 109–120 CrossRef PubMed 40 Chen, Y., Wang, K. and Zhu, D.H. (2006) Isolation, culture, identification and biological characteristics of chicken skeletal muscle satellite cells. Yi Chuan 28, 257–260 CrossRef PubMed 41 Rodrigues, A.M., Christen, B., Marti, M. and Izpisua Belmonte, J.C. (2012) Skeletal muscle regeneration in Xenopus tadpoles and zebrafish larvae. BMC Dev. Biol. 12, 9 CrossRef PubMed 42 Doherty, J.R., Johnson Hamlet, M.R., Kuliyev, E. and Mead, P.E. (2007) A flk-1 promoter/enhancer reporter transgenic Xenopus laevis generated using the Sleeping Beauty transposon system: an in vivo model for vascular studies. Dev. Dyn. 236, 2808–2817 CrossRef PubMed 43 Taylor, A.J. and Beck, C.W. (2012) Histone deacetylases are required for amphibian tail and limb regeneration but not development. Mech. Dev. 129, 208–218 CrossRef PubMed 44 Sugiura, T., Taniguchi, Y., Tazaki, A., Ueno, N., Watanabe, K. and Mochii, M. (2004) Differential gene expression between the embryonic tail bud and regenerating larval tail in Xenopus laevis. Dev. Growth Differ. 46, 97–105 CrossRef PubMed  C The

C 2014 Biochemical Society Authors Journal compilation 

45 Christen, B., Beck, C.W., Lombardo, A. and Slack, J.M. (2003) Regeneration-specific expression pattern of three posterior Hox genes. Dev. Dyn. 226, 349–355 CrossRef PubMed 46 Love, N.R., Chen, Y., Ishibashi, S., Kritsiligkou, P., Lea, R., Koh, Y., Gallop, J.L., Dorey, K. and Amaya, E. (2013) Amputation-induced reactive oxygen species are required for successful Xenopus tadpole tail regeneration. Nat. Cell Biol. 15, 222–228 CrossRef PubMed 47 Finkel, T. and Holbrook, N.J. (2000) Oxidants, oxidative stress and the biology of ageing. Nature 408, 239–247 CrossRef PubMed 48 Lambeth, J.D. (2004) NOX enzymes and the biology of reactive oxygen. Nat. Rev. Immunol. 4, 181–189 CrossRef PubMed 49 Denayer, T., Tran, H.T. and Vleminckx, K. (2008) Transgenic reporter tools tracing endogenous canonical Wnt signaling in Xenopus. Methods Mol. Biol. 469, 381–400 CrossRef PubMed 50 Funato, Y., Michiue, T., Asashima, M. and Miki, H. (2006) The thioredoxin-related redox-regulating protein nucleoredoxin inhibits Wnt–β-catenin signalling through dishevelled. Nat. Cell Biol. 8, 501–508 CrossRef PubMed 51 Poss, K.D., Shen, J., Nechiporuk, A., McMahon, G., Thisse, B., Thisse, C. and Keating, M.T. (2000) Roles for Fgf signaling during zebrafish fin regeneration. Dev. Biol. 222, 347–358 CrossRef PubMed 52 Schmidt, A.J. and Weidman, T. (1964) Dehydrogenases and aldolase in the regenerating forelimb of the adult newt, Diemictylus viridescens. J. Exp. Zool. 155, 303–315 CrossRef PubMed 53 King, M.W., Nguyen, T., Calley, J., Harty, M.W., Muzinich, M.C., Mescher, A.L., Chalfant, C., N’Cho, M., McLeaster, K., McEntire, J. et al. (2003) Identification of genes expressed during Xenopus laevis limb regeneration by using subtractive hybridization. Dev. Dyn. 226, 398–409 CrossRef PubMed 54 Grow, M., Neff, A.W., Mescher, A.L. and King, M.W. (2006) Global analysis of gene expression in Xenopus hindlimbs during stage-dependent complete and incomplete regeneration. Dev. Dyn. 235, 2667–2685 CrossRef PubMed 55 Pearl, E.J., Barker, D., Day, R.C. and Beck, C.W. (2008) Identification of genes associated with regenerative success of Xenopus laevis hindlimbs. BMC Dev. Biol. 8, 66 CrossRef PubMed 56 Papandreou, I., Cairns, R.A., Fontana, L., Lim, A.L. and Denko, N.C. (2006) HIF-1 mediates adaptation to hypoxia by actively downregulating mitochondrial oxygen consumption. Cell Metab. 3, 187–197 CrossRef PubMed 57 Maxwell, P.H., Wiesener, M.S., Chang, G.W., Clifford, S.C., Vaux, E.C., Cockman, M.E., Wykoff, C.C., Pugh, C.W., Maher, E.R. and Ratcliffe, P.J. (1999) The tumour suppressor protein VHL targets hypoxia-inducible factors for oxygen-dependent proteolysis. Nature 399, 271–275 CrossRef PubMed 58 Pan, Y., Mansfield, K.D., Bertozzi, C.C., Rudenko, V., Chan, D.A., Giaccia, A.J. and Simon, M.C. (2007) Multiple factors affecting cellular redox status and energy metabolism modulate hypoxia-inducible factor prolyl hydroxylase activity in vivo and in vitro. Mol. Cell. Biol. 27, 912–925 CrossRef PubMed 59 Chandel, N.S., Maltepe, E., Goldwasser, E., Mathieu, C.E., Simon, M.C. and Schumacker, P.T. (1998) Mitochondrial reactive oxygen species trigger hypoxia-induced transcription. Proc. Natl. Acad. Sci. U.S.A. 95, 11715–11720 CrossRef PubMed 60 Chandel, N.S., McClintock, D.S., Feliciano, C.E., Wood, T.M., Melendez, J.A., Rodriguez, A.M. and Schumacker, P.T. (2000) Reactive oxygen species generated at mitochondrial complex III stabilize hypoxia-inducible factor-1α during hypoxia: a mechanism of O2 sensing. J. Biol. Chem. 275, 25130–25138 CrossRef PubMed 61 Kim, J.W., Tchernyshyov, I., Semenza, G.L. and Dang, C.V. (2006) HIF-1-mediated expression of pyruvate dehydrogenase kinase: a metabolic switch required for cellular adaptation to hypoxia. Cell Metab. 3, 177–185 CrossRef PubMed 62 Semenza, G.L., Jiang, B.H., Leung, S.W., Passantino, R., Concordet, J.P., Maire, P. and Giallongo, A. (1996) Hypoxia response elements in the aldolase A, enolase 1, and lactate dehydrogenase A gene promoters contain essential binding sites for hypoxia-inducible factor 1. J. Biol. Chem. 271, 32529–32537 CrossRef PubMed 63 Iyer, N.V., Kotch, L.E., Agani, F., Leung, S.W., Laughner, E., Wenger, R.H., Gassmann, M., Gearhart, J.D., Lawler, A.M., Yu, A.Y. et al. (1998) Cellular and developmental control of O2 homeostasis by hypoxia-inducible factor 1α. Genes Dev. 12, 149–162 CrossRef PubMed

Biochemical Society Annual Symposium No. 81: Biochemical Determinants of Tissue Regeneration

64 Takagi, H., King, G.L. and Aiello, L.P. (1998) Hypoxia upregulates glucose transport activity through an adenosine-mediated increase of GLUT1 expression in retinal capillary endothelial cells. Diabetes 47, 1480–1488 CrossRef PubMed 65 Denko, N.C. (2008) Hypoxia, HIF1 and glucose metabolism in the solid tumour. Nat. Rev. Cancer 8, 705–713 CrossRef PubMed 66 Greer, S.N., Metcalf, J.L., Wang, Y. and Ohh, M. (2012) The updated biology of hypoxia-inducible factor. EMBO J. 31, 2448–2460 CrossRef PubMed 67 Wheaton, W.W. and Chandel, N.S. (2011) Hypoxia. 2. Hypoxia regulates cellular metabolism. Am. J. Physiol. Cell Physiol. 300, C385–C393 CrossRef PubMed 68 Noguchi, T., Inoue, H. and Tanaka, T. (1986) The M1- and M2-type isozymes of rat pyruvate kinase are produced from the same gene by alternative RNA splicing. J. Biol. Chem. 261, 13807–13812 PubMed 69 Luo, W., Hu, H., Chang, R., Zhong, J., Knabel, M., O’Meally, R., Cole, R.N., Pandey, A. and Semenza, G.L. (2011) Pyruvate kinase M2 is a PHD3-stimulated coactivator for hypoxia-inducible factor 1. Cell 145, 732–744 CrossRef PubMed

70 Mazurek, S., Boschek, C.B., Hugo, F. and Eigenbrodt, E. (2005) Pyruvate kinase type M2 and its role in tumor growth and spreading. Semin. Cancer Biol. 15, 300–308 CrossRef PubMed 71 Christofk, H.R., Vander Heiden, M.G., Wu, N., Asara, J.M. and Cantley, L.C. (2008) Pyruvate kinase M2 is a phosphotyrosine-binding protein. Nature 452, 181–186 CrossRef PubMed 72 Anastasiou, D., Poulogiannis, G., Asara, J.M., Boxer, M.B., Jiang, J.K., Shen, M., Bellinger, G., Sasaki, A.T., Locasale, J.W., Auld, D.S. et al. (2011) Inhibition of pyruvate kinase M2 by reactive oxygen species contributes to cellular antioxidant responses. Science 334, 1278–1283 CrossRef PubMed

Received 14 March 2014 doi:10.1042/BST20140061

 C The

C 2014 Biochemical Society Authors Journal compilation 

623

Tadpole tail regeneration in Xenopus.

Some organisms have a remarkable ability to heal wounds without scars and to regenerate complex tissues following injury. By gaining a more complete u...
492KB Sizes 32 Downloads 4 Views